EP2054067A2 - Administration of high potency platinum compound formulations by inhalation - Google Patents

Administration of high potency platinum compound formulations by inhalation

Info

Publication number
EP2054067A2
EP2054067A2 EP07814117A EP07814117A EP2054067A2 EP 2054067 A2 EP2054067 A2 EP 2054067A2 EP 07814117 A EP07814117 A EP 07814117A EP 07814117 A EP07814117 A EP 07814117A EP 2054067 A2 EP2054067 A2 EP 2054067A2
Authority
EP
European Patent Office
Prior art keywords
lipid
platinum compound
compound formulation
platinum
soy
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP07814117A
Other languages
German (de)
English (en)
French (fr)
Inventor
Frank G. Pilkiewicz
Walter R. Perkins
Beth Metzheiser
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Transave LLC
Original Assignee
Transave LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Transave LLC filed Critical Transave LLC
Publication of EP2054067A2 publication Critical patent/EP2054067A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0078Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a nebulizer such as a jet nebulizer, ultrasonic nebulizer, e.g. in the form of aqueous drug solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes

Definitions

  • the present invention relates to a method for treating cancer by delivering a therapeutically effective amount of a lipid composition containing a cytotoxic agent (e.g., cisplatin) to a patient's respiratory tract.
  • a cytotoxic agent e.g., cisplatin
  • the method allows clinicians to administer treatment cycles more frequently without the attendant side effects (e.g., nephrotoxicity, bone marrow toxicity) common to systemic administration of many cancer cytotoxic agents (e.g., cisplatin).
  • Bronchoalveolar Carcinoma or alveolar cell carcinoma is a form of adenocarcinoma, a cell-type of non-small cell carcinoma of the lung which can be found throughout the respiratory tract.
  • BAC represents approximately 10 to 25% of the adenocarcinoma of lung cases or 2-6% of all lung cancers and sometimes has a distinct presentation and biologic behavior.
  • BAC is more common in women and in patients who do not smoke cigarettes than other histologic types of lung cancer.
  • BAC may present as a solitary peripheral nodule, a multifocal lesion, or a rapidly progressive form that appears as a diffuse infiltrate on chest radiograph.
  • the cells secrete mucin and surfactant apoprotein which can lead to bronchorrhea, an excessive discharge of mucus from the air passages of the lungs.
  • Bronchoalveolar cancer may present as a more diffuse lesion than other types of cancer. When it is discovered as a single mass on a patient's x-ray, this type of lung cancer has an excellent prognosis. Five year survival after surgery is in the 75-90 percent range. If, however, it is found in its diffuse form (meaning it has spread beyond a single mass), the prognosis is quite poor.
  • the management and prognosis are essentially the same as other types of non-small cell lung cancer.
  • Surgery is the preferred treatment if the tumor can be resected. Radiation therapy and chemotherapy may be used in non-operable cases. Trials are underway to investigate treatments specific for bronchoalveolar carcinoma.
  • Carcinomatosis with lymphangitic spread, or Lymphangitis carcinomatosis refers to the diffuse infiltration and obstruction of pulmonary parenchymal lymphatic channels by tumor.
  • Various neoplasms can cause lymphangitic carcinomatosis, but 80% are adenocarcinomas. The most frequent primary sites are the breast, lungs, colon, and stomach.
  • Other sources include the pancreas, thyroid, cervix, prostate, larynx, and metastatic adenocarcinoma from an unknown primary.
  • LC occurs as a result of initial hematogenous spread of tumor to the lungs, with subsequent malignant invasion through the vessel wall into the pulmonary interstitium and lymphatics. The tumor then proliferates and spreads easily through these low resistance channels. Less commonly, direct infiltration occurs from contiguous mediastinal or hilar lymphadenopathy or from an adjacent primary bronchogenic carcinoma. Histopatho logically, interstitial edema, interstitial fibrosis (secondary to a desmoplastic reaction as a result of tumor extension into adjacent pulmonary parenchyma), and tumor cells all can be seen. Metastatic adenocarcinoma accounts for 80% of cases. Most patients are middle-aged adults.
  • LC represents 7% of all pulmonary metastases. Prevalence in postmortem studies is significantly higher than the incidence of radiologically detectable disease. Microscopic interstitial tumor invasion is seen in 56% of patients with pulmonary metastases. Prognosis for patients with LC is poor. Most patients survive only weeks or months.
  • chemo therapeutic treatment of lung cancers includes systemic administration of chemotherapeutic agents, e.g., cytotoxic agents, to the patients.
  • cisplatin cis-diamine-dichloroplatinum (H)
  • H cis-diamine-dichloroplatinum
  • nephrotoxicity limits the frequency in which clinicians can administer cisplatin to the patient.
  • successive treatment cycles of cisplatin typically require three weeks or more between treatment cycles to prevent blood levels of cisplatin from reaching those correlated with nephrotoxicity.
  • chemotherapeutic regimens typically require five or more treatment cycles, the delay between treatment cycles lengthens the time needed for the overall chemotherapeutic regimen.
  • the prolonged time periods for systemic administration of cisplatin lead to increased patient discomfort and inconvenience, and may lead to decreased patient compliance.
  • Cisplatin is difficult to efficiently entrap in liposomes or lipid complexes because of the bioactive agent's low aqueous solubility, approximately 1.0 mg/ml at room temperature, and low lipophilicity, both of which properties contribute to a low bioactive agent/lipid ratio.
  • Liposomes and lipid complexes containing cisplatin suffer from another problem - stability of the composition.
  • maintenance of bioactive agent potency and retention of the bioactive agent in the liposome during storage are recognized problems (Fumble, et al., 1982; Gondal, et al., 1993; Potkul, et al., 1991 Am J Obstet Gynecol. 164(2): 652-658; Steerenberg, et al., 1988; Weiss, et al., 1993) and a limited shelf life of liposomes containing cisplatin, on the order of several weeks at 4° C, has been reported (Gondal, et al., 1993 Eur J Cancer. 29A(I l): 1536-1542; Potkul, et al., 1991).
  • the present invention results from the realization that effective treatment with reduced administration times can be achieved with high potency platinum compound formulations.
  • high potency it is meant that the concentration of the platinum compound is higher than its aqueous solubility.
  • cisplatin high potency would be concentrations higher than its aqueous solubility of 1 mg/ml.
  • Platinol® for example, is a commercial cisplatin product by Bristol Meyers Squibb having 1 mg/ml cisplatin.
  • the present invention relates to a method of treating cancer or a method of reducing treatment times for a patient comprising administering to the patient by inhalation, a cancer treating effective amount of a lipid-based platinum compound formulation wherein the concentration of the platinum compound of the lipid-based platinum compound formulation is greater than its aqueous solubility.
  • the platinum compound concentration is 1.2 mg/ml, 3 mg/ml, or 5 mg/ml.
  • the platinum compound is selected from the group consisting of: cisplatin, carboplatin (diammine(l, 1 -cyclobutanedicarboxylato)-platinum(II)), tetraplatin (ormaplatin) (tetrachloro(l ,2-cyclohexanediamine-N,N')-platinum(IV)), thioplatin (bis(O- ethyldithiocarbonato)platinum(II)), satraplatin, nedaplatin, DACH-platinum such as oxaliplatin and aroplatin, heptaplatin, iproplatin, transplatin, lobaplatin, cis- aminedichloro(2-methylpyridine) platinum, JMl 18 (czs-amminedichloro (cyclohexylamine)platinum(II)), JM 149 (c ⁇ -amminedichloro(cyclohexyl
  • the platinum compound is cisplatin.
  • the lipids used in the present invention can be synthetic, semi-synthetic or naturally-occurring lipids, including phospholipids, tocopherols, sterols, fatty acids, glycolipids, anionic lipids, cationic lipids.
  • phosho lipids they can include such lipids as egg phosphatidylcholine (EPC), egg phosphatidylglycerol (EPG), egg phosphatidylinositol (EPI), egg phosphatidylserine (EPS), phosphatidylethanolamine (EPE), and phosphatidic acid (EPA); the soya counterparts, soy phosphatidylcholine (SPC); SPG, SPS, SPI, SPE, and SPA; the hydrogenated egg and soya counterparts (e.g., HEPC, HSPC), sterically modified phosphatidylethanolamines, cholesterol derivatives, carotinoids, other phospholipids made up of ester linkages of fatty acids in the 2 and 3 of glycerol positions containing chains of 12 to 26 carbon atoms and different head groups in the 1 position of glycerol that include choline, glycerol, inositol, serine,
  • the chains on these fatty acids can be saturated or unsaturated, and the phospholipid may be made up of fatty acids of different chain lengths and different degrees of unsaturation.
  • the compositions of the formulations can include DPPC, a major constituent of naturally-occurring lung surfactant.
  • DMPC dimyristoylphosphatidycholine
  • DMPG dimyristoylphosphatidylg- lycerol
  • DPPC dipalmitoylphosphatidcholine
  • DPPG dipalmitoylphosphatidylglycerol
  • DSPC distearoylphosphatidylcholine
  • DSPG distearoylphosphatidylglycerol
  • DOPE dioleylphosphatidyl-ethanolamine
  • PSPC palmitoylstearoylphosphatidylcholine
  • PSPG palmitoylstearolphosphatidylglycerol
  • triacylglycerol diacylglycerol
  • sphingosine ceramide
  • sphingomyelin single acylated phospholipids like mono-oleoyl-phosphatidylethanolarnine (MOPE), and quadruple acylated.
  • the lipid in the lipid-based platinum compound formulation is a mixture of a phospholipid and a sterol, such as for example, DPPC and cholesterol.
  • the lipid in the lipid-based platinum compound formulation is a mixture of DPPC from 50 to 65 mol % and cholesterol from 35 to 50 mol%.
  • the cancer in one embodiment, may be lung cancer, such as, for example, BAC or LC.
  • the ratio of platinum compound to lipid in the lipid- based platinum compound formulation is between 1 :5 by weight and 1 :50 by weight, hi still a another embodiment, the lipid-based platinum compound formulation comprises liposomes having a mean diameter of 0.01 microns to 3.0 microns.
  • the lipid is a mixture of DPPC and cholesterol, the ratio of platinum compound to lipid in the lipid-based platinum compound formulation is between 1 :5 by weight and 1 :50 by weight, and wherein the lipid-based platinum compound formulation comprises liposomes having a mean diameter of 0.01 microns to 3.0 microns.
  • the lipid is a mixture of DPPC and cholesterol, the ratio of platinum compound to lipid in the lipid-based platinum compound formulation is between 1 :5 by weight and 1 : 50 by weight, the lipid-based platinum compound formulation comprises liposomes having a mean diameter of 0.01 microns to 3.0 microns, and wherein the platinum compound is cisplatin.
  • the lipid is a mixture of DPPC and cholesterol in a 2 to 1 ratio by weight, the ratio of platinum compound to lipid in the lipid-based platinum compound formulation is 1 :20 by weight, the lipid-based platinum compound formulation comprises liposomes having a mean diameter of 0.40 microns, and wherein the platinum compound is cisplatin.
  • the patient is preferably a human.
  • the lipid-based platinum compound formulation is administered to the patient at least once every three weeks.
  • the amount of platinum compound in the lipid-based platinum compound formulation is 18 mg/m or greater, 24 mg/m or greater, 36 mg/m or greater, or 48 mg/m 2 or greater.
  • the amount of platinum compound in the lipid-based platinum compound formulation is 100 mg/m 2 or greater, and the lipid-based platinum compound formulation is administered to the patient at least once every three weeks, and preferably once every two weeks.
  • Figure 1 depicts the aerodynamic particle size distributions of liposomal cisplatin lots at different potencies: 1 mg/ml and 5 mg/ml.
  • Figure 2 depicts Pt levels in the lungs of rats as a function of time after inhalation. Whole lungs were homogenized. Points represent the mean of three samples ⁇ s.d.
  • Figure 3 depicts Pt levels in the lymph nodes of rats as a function of time after inhalation. Whole bronchial lymph nodes were homogenized. Points represent the mean of three samples ⁇ s.d.
  • platinum compound is a compound containing coordinated platinum and having antineoplastic activity.
  • Active platinum compounds include, for example, cisplatin, carboplatin, and DACH-platinum compounds such as oxaplatin and aroplatin.
  • a "patient,” “subject” or “host” to be treated by the subject method may mean either a human or non-human animal.
  • therapeutic effect is art-recognized and refers to a local or systemic effect in animals, particularly mammals, and more particularly humans caused by a pharmacologically active substance.
  • therapeutically-effective amount means that amount of a substance that produces some desired local or systemic effect at a reasonable benefit/risk ratio applicable to any treatment.
  • the therapeutically effective amount of a substance will vary depending upon the subject and disease condition being treated, the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • treating is art-recognized and refers to curing as well as ameliorating at least one symptom of a condition or disease or preventing the occurrence of a condition or disease.
  • Treatment cycle means the time period in which a given dose of cisplatin is to be administered to a patient.
  • Treatment cycles may encompass one or more sessions where the patient is actively being administered the lipid composition containing cisplatin. Such sessions may be administered over the course of four days or less, but more preferably is administered over one or two days, and most preferably is administered in one day.
  • High potency lipid formulations of platinum compounds have been designed for aerosol administration to patients with primary and metastatic cancer in the lungs. Aerosolized platinum compound formulations enable effective targeting of the drug to the lungs without significant systemic levels. The improved lung targeting reduces systemic toxicity and enables larger doses of therapeutic to be delivered.
  • a critical success factor for administering a clinically therapeutic dose of a platinum compound formulation would be to do so in no more than 1 hr. Preferably, this would be accomplished without a formulation or device change.
  • One potential way to do this is to increase the potency of the drug product (i.e., reduce water content from the product).
  • the aerodynamic particle size distribution of a more concentrated formulation of cisplatin, Lot B (5 mg/ml cisplatin), in comparison with Lot A (1 mg/ml cisplatin) has been examined. Provided the aerosol characteristics of the two products are equivalent, the five-fold more potency would enable doses up to 24 mg/m 2 to be delivered in less than the 1 hr target (ca., 32 min).
  • the mass median aerodynamic diameters (MMAD) and geometric standard deviations (GSD) for the formulations are presented in Table 1.
  • the MMAD values range from about 3.3 ⁇ m to 3.9 ⁇ m.
  • the mean GSD values for the 1 mg/ml and 5 mg/ml formulations are 1.94 and 1.89, respectively.
  • the rates of nebulization for the formulations are presented in Table 2.
  • the rate of nebulization range from about 0.26 g/min to 0.39 g/min.
  • Increasing the potency of liposomal cisplatin from 1 mg/ml to 5 mg/ml does not result in significant differences in the aerodynamic particle size distribution of the nebulized droplets.
  • Increasing the potency of liposomal cisplatin from 1 mg/ml to 5 mg/ml does not result in significant differences in the rate of nebulization. Hence, it is anticipated that the increase in potency will enable five-fold reductions in administration time.
  • the lipids used in the present invention can be synthetic, semi-synthetic or naturally-occurring lipids, including phospholipids, tocopherols, sterols, fatty acids, glycolipids, anionic lipids, cationic lipids.
  • phosho lipids they can include such lipids as egg phosphatidylcholine (EPC), egg phosphatidylglycerol (EPG), egg phosphatidylinositol (EPI), egg phosphatidylserine (EPS), phosphatidylethanolamine (EPE), and phosphatidic acid (EPA); the soya counterparts, soy phosphatidylcholine (SPC); SPG, SPS, SPI, SPE, and SPA; the hydrogenated egg and soya counterparts (e.g., HEPC, HSPC), sterically modified phosphatidylethanolamines, cholesterol derivatives, carotinoids, other phospholipids made up of ester linkages of fatty acids in the 2 and 3 of glycerol positions containing chains of 12 to 26 carbon atoms and different head groups in the 1 position of glycerol that include choline, glycerol, inositol, serine,
  • the chains on these fatty acids can be saturated or unsaturated, and the phospholipid may be made up of fatty acids of different chain lengths and different degrees of unsaturation.
  • the compositions of the formulations can include DPPC, a major constituent of naturally-occurring lung surfactant.
  • DMPC dimyristoylphosphatidycholine
  • DMPG dimyristoylphosphatidylg- lycerol
  • DPPC dipalmitoylphosphatidcholine
  • DPPG dipalmitoylphosphatidylglycerol
  • DSPC distearoylphosphatidylcholine
  • DSPG distearoylphosphatidylglycerol
  • DOPE dioleylphosphatidyl-ethanolamine
  • PSPC palmitoylstearoylphosphatidylcholine
  • PSPG palmitoylstearolphosphatidylglycerol
  • triacylglycerol diacylglycerol
  • sphingosine ceramide
  • sphingomyelin single acylated phospholipids like mono-oleoyl-phosphatidylethanolarnine (MOPE), and quadruple acylated.
  • the sterols can include, cholesterol, esters of cholesterol including cholesterol hemi- succinate, salts of cholesterol including cholesterol hydrogen sulfate and cholesterol sulfate, ergosterol, esters of ergosterol including ergosterol hemi-succinate, salts of ergosterol including ergosterol hydrogen sulfate and ergosterol sulfate, lanosterol, esters of lanosterol including lanosterol hemi-succinate, salts of lanosterol including lanosterol hydrogen sulfate and lanosterol sulfate.
  • the lipid composition contains 50 to 100 mol% DPPC and 0 to 50 mol% cholesterol. More preferably, the lipid complex contains 50 to 65 mol% DPPC and 35 to 50 mol% cholesterol.
  • the inhalation delivery device of the inhalation system can be a nebulizer, a metered dose inhaler (MDI) or a dry powder inhaler (DPI).
  • MDI metered dose inhaler
  • DPI dry powder inhaler
  • the device can contain and be used to deliver a single dose of the lipid compositions or the device can contain and be used to deliver multi-doses of the lipid compositions of the present invention.
  • a nebulizer type inhalation delivery device can contain the compositions of the present invention as a solution, usually aqueous, or a suspension.
  • the nebulizer type delivery device may be driven ultrasonically, by compressed air, by other gases, electronically or mechanically (including, for example, a vibrating porous membrane).
  • the ultrasonic nebulizer device usually works by imposing a rapidly oscillating waveform onto the liquid film of the formulation via an electrochemical vibrating surface. At a given amplitude the waveform becomes unstable, whereby it disintegrates the liquids film, and it produces small droplets of the formulation.
  • the nebulizer device driven by air or other gases operates on the basis that a high pressure gas stream produces a local pressure drop that draws the liquid formulation into the stream of gases via capillary action. This fine liquid stream is then disintegrated by shear forces.
  • the nebulizer may be portable and hand held in design, and may be equipped with a self contained electrical unit.
  • the nebulizer device can consist of a nozzle that has two coincident outlet channels of defined aperture size through which the liquid formulation can be accelerated. This results in impaction of the two streams and atomization of the formulation.
  • the nebulizer may use a mechanical actuator to force the liquid formulation through a multiorifice nozzle of defined aperture size(s) to produce an aerosol of the formulation for inhalation.
  • blister packs containing single doses of the formulation may be employed.
  • the nebulizer is employed to ensure the sizing of aqueous droplets containing the drug-lipid particles is optimal for positioning of the particle within, for example, the lungs.
  • Typical droplet sizes for the nebulized lipid composition are from 1 to 5 microns.
  • the lipid composition preferably contains an aqueous component. Typically there is at least 80% by weight and preferably, at least 90% by weight of the aqueous component in the lipid composition to be administered with a nebulizer.
  • the aqueous component may include for example, saline.
  • the aqueous component may include up to 20% by weight of an aqueous compatible solvent such as ethanol.
  • Total administration time using a nebulizer will depend on the flow rate and the concentration of the cisplatin in the lipid composition. Variation of the total administration time is within the purview of those of ordinary skill in the art. Generally, the flow rate of the nebulizer will be at least 0.15 ml/min, for example, a flow rate of 0.2 ml/min is typical.
  • administration of a dose of 36 mg/m 2 of cisplatin using a lipid composition having a concentration of 1 mg/ml of cisplatin would be 4 hours (assuming a patient's body surface area is 2 m 2 ).
  • This administration time may, for example, be split into two administration sessions given over the course of one or two days to complete one treatment cycle. More preferably, administration of a dose of 36 mg/m 2 of cisplatin using a lipid composition having a concentration of 5 mg/ml of cisplatin would be 48 minutes to complete one treatment cycle.
  • a metered dose inhaler can be employed as the inhalation delivery device of the inhalation system.
  • This device is pressurized (pMDI) and its basic structure consists of a metering valve, an actuator and a container.
  • a propellant is used to discharge the formulation from the device.
  • the composition can consist of particles of a defined size suspended in the pressurized propellant(s) liquid, or the composition can be in a solution or suspension of pressurized liquid propellant(s).
  • the propellants used are primarily atmospheric friendly hydro flourocarbons (HFCs) such as 134a and 227. Traditional chloro flourocarbons like CFC-I 1, 12 and 114 are used only when essential.
  • HFCs atmospheric friendly hydro flourocarbons
  • the device of the inhalation system may deliver a single dose via, e.g., a blister pack, or it may be multi dose in design.
  • the pressurized metered dose inhaler of the inhalation system can be breath actuated to deliver an accurate dose of the lipid based formulation.
  • the delivery of the formulation may be programmed via a microprocessor to occur at a certain point in the inhalation cycle.
  • the MDI may be portable and hand held.
  • a dry powder inhaler (DPI) can be used as the inhalation delivery device of the inhalation system.
  • This device's basic design consists of a metering system, a powdered composition and a method to disperse the composition.
  • the metering and dispersion systems may be mechanically or electrically driven and may be microprocessor programmable.
  • the device may be portable and hand held.
  • the inhaler may be multi or single dose in design and use such options as hard gelatin capsules, and blister packages for accurate unit doses.
  • the composition can be dispersed from the device by passive inhalation; i.e., the patient's own inspiratory effort, or an active dispersion system may be employed.
  • the dry powder of the composition can be sized via processes such as jet milling, spray dying and supercritical fluid manufacture. Acceptable excipients such as the sugars mannitol and maltose may be used in the preparation of the powdered formulations.
  • sugars help in maintaining the liposome's physical characteristics during freeze drying and minimizing their aggregation when they are administered by inhalation.
  • the hydroxyl groups of the sugar may help the vesicles maintain their tertiary hydrated state and help minimize particle aggregation.
  • the inventive method is particularly well-suited for the treatment of lung cancers, particularly, bronchoalveolar carcinoma, or carcinomatosis with lymphangitic spread, hi addition, both primary and metastatic lung cancers are excellent candidates for the method of the invention.
  • any composition of the present invention will vary depending on the symptoms, age and body weight of the patient, the nature and severity of the disorder to be treated or prevented, the route of administration, and the form of the supplement. Any of the subject formulations may be administered in a single dose or in divided doses. Dosages for the compounds of the present invention may be readily determined by techniques known to those of skill in the art or as taught herein. Also, the present invention contemplates mixtures of more than one subject compound, as well as other therapeutic agents. Further, the present invention contemplates administration of the therapeutic agent that is contained in a subject coordination complex (or a related agent) in conjunction with the complex itself to increase the ratio of the therapeutic agent to the coordination complex formed upon release of the therapeutic agent,
  • the dosage of the subject compounds will generally be in the range of 0.01 ng to 1O g per kg body weight, specifically in the range of 1 ng to 0.1 g per kg, and more specifically in the range of 100 ng to 10 mg per kg.
  • An effective dose or amount, and any possible affects on the timing of administration of the formulation may need to be identified for any particular compound of the present invention. This may be accomplished by routine experiment as described herein, using one or more groups of animals (preferably at least 5 animals per group), or in human trials if appropriate.
  • the effectiveness of any compound and method of treatment or prevention may be assessed by administering the supplement and assessing the effect of the administration by measuring one or more indices associated with the neoplasm of interest, and comparing the post-treatment values of these indices to the values of the same indices prior to treatment.
  • the precise time of administration and amount of any particular compound that will yield the most effective treatment in a given patient will depend upon the activity, pharmacokinetics, and bioavailability of a particular compound, physiological condition of the patient (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage and type of medication), route of administration, and the like.
  • the guidelines presented herein may be used to optimize the treatment, e.g., determining the optimum time and/or amount of administration, which will require no more than routine experimentation consisting of monitoring the subject and adjusting the dosage and/or timing.
  • the health of the patient may be monitored by measuring one or more of the relevant indices at predetermined times during a 24-hour period. Treatment, including supplement, amounts, times of administration and formulation, may be optimized according to the results of such monitoring.
  • the patient may be periodically reevaluated to determine the extent of improvement by measuring the same parameters, the first such revaluation typically occurring at the end of four weeks from the onset of therapy, and subsequent reevaluations occurring every four to eight weeks during therapy and then every three months thereafter. Therapy may continue for several months or even years, with a minimum of one month being a typical length of therapy for humans. Adjustments to the amount(s) of agent administered and possibly to the time of administration may be made based on these reevaluations.
  • Treatment may be initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage may be increased by small increments until the optimum therapeutic effect is attained.
  • the combined use of several compounds of the present invention, or alternatively other chemotherapeutic agents, may reduce the required dosage for any individual component because the onset and duration of effect of the different components may be complimentary.
  • the different active agents may be delivered together or separately, and simultaneously or at different times within the day.
  • Toxicity and therapeutic efficacy of subject compounds may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 and the ED 50 .
  • Compositions that exhibit large therapeutic indices are preferred. Although compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets the compounds to the desired site in order to reduce side effects.
  • the data obtained from the cell culture assays and animal studies may be used in formulating a range of dosage for use in humans.
  • the dosage of any supplement, or alternatively of any components therein lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose may be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC 50 i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • Kits This invention also provides kits for conveniently and effectively implementing the methods of this invention.
  • kits comprise any of the compounds of the present invention or a combination thereof, and a means for facilitating compliance with methods of this invention.
  • kits provide a convenient and effective means for assuring that the subject to be treated takes the appropriate active in the correct dosage in the correct manner.
  • the compliance means of such kits includes any means which facilitates administering the actives according to a method of this invention.
  • Such compliance means include instructions, packaging, and dispensing means, and combinations thereof. Kit components may be packaged for either manual or partially or wholly automated practice of the foregoing methods. In other embodiments involving kits, this invention contemplates a kit including compositions of the present invention, and optionally instructions for their use.
  • the following examples further illustrate the present invention, but of course, should not be construed as in any way limiting its scope.
  • Step 3) is repeated 2-3 times.
  • the dispersion After washing process, the dispersion provides 1 mg/ml cisplatin potency and concentrated to 3 mg/ml cisplatin and further concentrated to 5 mg/ml cisplatin by aseptically removing two third of the aqueous vehicle of 1 mg/ml product and four fifth of the aqueous vehicle of 1 mg/ml product, respectively.
  • the removal of aqueous vehicle was carried out at a rate of 100 ml/min by diafiltration at 20 0 C without compensating the permeate with fresh sterile 0.9% sodium chloride solution.
  • *total duration includes rest time between actual dose administration(5min between each nebulization and 1 hour after every three nebulizations).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Inorganic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Dispersion Chemistry (AREA)
  • Otolaryngology (AREA)
  • Pulmonology (AREA)
  • Immunology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
EP07814117A 2006-08-16 2007-08-15 Administration of high potency platinum compound formulations by inhalation Withdrawn EP2054067A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11/505,236 US20070065522A1 (en) 2004-03-18 2006-08-16 Administration of high potency platinum compound formulations by inhalation
PCT/US2007/075995 WO2008022189A2 (en) 2006-08-16 2007-08-15 Administration of high potency platinum compound formulations by inhalation

Publications (1)

Publication Number Publication Date
EP2054067A2 true EP2054067A2 (en) 2009-05-06

Family

ID=39083094

Family Applications (1)

Application Number Title Priority Date Filing Date
EP07814117A Withdrawn EP2054067A2 (en) 2006-08-16 2007-08-15 Administration of high potency platinum compound formulations by inhalation

Country Status (6)

Country Link
US (1) US20070065522A1 (ja)
EP (1) EP2054067A2 (ja)
JP (1) JP2010501006A (ja)
CA (1) CA2660229A1 (ja)
MX (1) MX2009001542A (ja)
WO (1) WO2008022189A2 (ja)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9107824B2 (en) 2005-11-08 2015-08-18 Insmed Incorporated Methods of treating cancer with high potency lipid-based platinum compound formulations administered intraperitoneally
US8173686B2 (en) 2006-11-06 2012-05-08 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8168662B1 (en) 2006-11-06 2012-05-01 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8168661B2 (en) * 2006-11-06 2012-05-01 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8178564B2 (en) * 2006-11-06 2012-05-15 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US20110033528A1 (en) * 2009-08-05 2011-02-10 Poniard Pharmaceuticals, Inc. Stabilized picoplatin oral dosage form
TW200916094A (en) * 2007-06-27 2009-04-16 Poniard Pharmaceuticals Inc Stabilized picoplatin dosage form
US20100260832A1 (en) * 2007-06-27 2010-10-14 Poniard Pharmaceuticals, Inc. Combination therapy for ovarian cancer
EP2178893A4 (en) * 2007-07-16 2012-09-19 Poniard Pharmaceuticals Inc ORAL FORMULATIONS FOR PICOPLATIN
CN102006875A (zh) * 2008-02-08 2011-04-06 帕纳德制药公司 吡铂和贝伐单抗治疗结直肠癌的用途
WO2011005377A2 (en) * 2009-05-27 2011-01-13 Mount Sinai School Of Medicine Of New York University Compositions and methods comprising vegfr-2 and vegfr-3 antagonists for the treatment of metastatic disease
MX369543B (es) * 2011-06-17 2019-11-12 Berg Llc Composiciones farmaceuticas inhalables.
CN112451486A (zh) 2012-09-04 2021-03-09 埃莱森制药有限责任公司 用顺铂脂质复合物预防癌症的肺部复发
WO2015092123A1 (en) * 2013-12-18 2015-06-25 Simo Rasi Compositions comprising glycerophosphorylethanolamine
US20190307690A1 (en) * 2016-05-03 2019-10-10 The American University In Cairo Liposomal Delivery Systems for Oxaliplatin and in Dual Drug Delivery in Combination with Chemo-sensitizing and Chemo-therapeutic agents
WO2020135920A1 (en) * 2018-12-28 2020-07-02 Université Libre de Bruxelles Kit for inhaled chemotherapy, and treatment of lung cancer with said kit

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4981692A (en) * 1983-03-24 1991-01-01 The Liposome Company, Inc. Therapeutic treatment by intramammary infusion
US4590001A (en) * 1983-03-28 1986-05-20 Stjernholm Rune L Platinum bound to transferrin for use in the treatment of breast tumors
US5019369A (en) * 1984-10-22 1991-05-28 Vestar, Inc. Method of targeting tumors in humans
US5041581A (en) * 1985-10-18 1991-08-20 The University Of Texas System Board Of Regents Hydrophobic cis-platinum complexes efficiently incorporated into liposomes
US5049388A (en) * 1986-11-06 1991-09-17 Research Development Foundation Small particle aerosol liposome and liposome-drug combinations for medical use
US5320906A (en) * 1986-12-15 1994-06-14 Vestar, Inc. Delivery vehicles with amphiphile-associated active ingredient
US5616334A (en) * 1987-03-05 1997-04-01 The Liposome Company, Inc. Low toxicity drug-lipid systems
IL83380A (en) * 1987-07-30 1991-04-15 Teva Pharma Stable aqueous cisplatin solutions
CA1335348C (en) * 1988-03-04 1995-04-25 Yasuaki Ogawa Liposome composition
US5141751A (en) * 1988-06-29 1992-08-25 Daiichi Pharmaceutical Co., Ltd. Lipid membrane structures
US5013556A (en) * 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5858784A (en) * 1991-12-17 1999-01-12 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol- and liposome-based delivery
US5756353A (en) * 1991-12-17 1998-05-26 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol-and liposome-based delivery
US5958449A (en) * 1992-12-02 1999-09-28 Nexstar Pharmaceuticals, Inc. Antibiotic formulation and use for bacterial infections
US5780054A (en) * 1996-01-17 1998-07-14 University Of British Columbia Methods for increasing the circulation half-life of protein-based therapeutics
US20010006648A1 (en) * 1996-02-26 2001-07-05 Hitoshi Yamauchi Liposomes and liposomal dispersion
CA2263455C (en) * 1996-08-23 2002-10-29 Sequus Pharmaceuticals, Inc. Liposomes containing a cisplatin compound
US5997899A (en) * 1996-10-01 1999-12-07 Skyepharma Inc. Method for producing liposomes with increased percent of compound encapsulated
US6451784B1 (en) * 1996-12-30 2002-09-17 Battellepharma, Inc. Formulation and method for treating neoplasms by inhalation
US6630168B1 (en) * 1997-02-20 2003-10-07 Biomedicines, Inc. Gel delivery vehicles for anticellular proliferative agents
US6787132B1 (en) * 1997-12-04 2004-09-07 Yissum Research Development Company Of The Hebrew University Of Jerusalem Combined chemo-immunotherapy with liposomal drugs and cytokines
US6726925B1 (en) * 1998-06-18 2004-04-27 Duke University Temperature-sensitive liposomal formulation
CA2248592A1 (en) * 1998-08-31 2000-02-29 Christopher D. Batich Microspheres for use in the treatment of cancer
US20050074499A1 (en) * 1999-03-17 2005-04-07 Mitsubishi Chemical Corporation Ligand-bonded complex
US6723338B1 (en) * 1999-04-01 2004-04-20 Inex Pharmaceuticals Corporation Compositions and methods for treating lymphoma
US6852334B1 (en) * 1999-04-20 2005-02-08 The University Of British Columbia Cationic peg-lipids and methods of use
EP1259225B1 (en) * 2000-02-04 2006-10-04 Lipoxen Technologies Limited Process of dehydration/rehydration for making liposomes
CA2412795C (en) * 2000-06-30 2012-10-02 Inex Pharmaceuticals Corporation Improved liposomal camptothecins and uses thereof
WO2002085386A2 (en) * 2001-04-23 2002-10-31 Nucryst Pharmaceuticals Corp. Medicament containing a metal such as silver, gold, platinum or palladium as an antimicrobial agent and their use to induce apoptosis in cancerous tissue
DE60210402T2 (de) * 2001-05-18 2006-12-07 Chiron Corp., Emeryville System zur Abgabe einer Tobramycin-Formulierung
US7063860B2 (en) * 2001-08-13 2006-06-20 University Of Pittsburgh Application of lipid vehicles and use for drug delivery
AU2002323266B2 (en) * 2001-08-20 2008-04-24 Transave, Inc. Method for treating lung cancers
NZ538179A (en) * 2002-08-02 2008-09-26 Transave Inc Liposome or lipid complex with active platinum compound and process for producing the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2008022189A2 *

Also Published As

Publication number Publication date
CA2660229A1 (en) 2008-02-21
MX2009001542A (es) 2009-03-20
WO2008022189A3 (en) 2008-11-13
JP2010501006A (ja) 2010-01-14
US20070065522A1 (en) 2007-03-22
WO2008022189A2 (en) 2008-02-21

Similar Documents

Publication Publication Date Title
US20070065522A1 (en) Administration of high potency platinum compound formulations by inhalation
US20050249822A1 (en) Administration of cisplatin by inhalation
AU2002323266B2 (en) Method for treating lung cancers
JP6357481B2 (ja) プロスタサイクリン化合物およびプロスタサイクリン化合物を使用する方法
Rudokas et al. Liposome delivery systems for inhalation: a critical review highlighting formulation issues and anticancer applications
AU2002323266A1 (en) Method for treating lung cancers
AU2013266400B2 (en) Systems for treating pulmonary infections
EP1962805B1 (en) Lipid-based compositions of antiinfectives for treating pulmonary infections
US20050107287A1 (en) Treatment of cancers by inhalation of stable platinum-containing formulations
US20060034906A1 (en) Treatment of lung diseases and pre-lung disease conditions
WO2015138423A9 (en) Prostacylin compositions and methods for using the same
BRPI0619475B1 (pt) Composições farmacêuticas na forma de lipossomas compreendendo ciclosporina a, seu método de preparo e usos
US20060159712A1 (en) Lipid particles comprising bioactive agents, methods of preparing and uses thereof
EP1811963A2 (en) Methods of treating cancer with lipid-based platinum compound formulations administered intraperitoneally
Chraibi et al. The combination of an innovative dry powder for inhalation and a standard cisplatin-based chemotherapy in view of therapeutic intensification against lung tumours
US20240252431A1 (en) Pulmonary biguanide formulations and delivery devices
CN117224482A (zh) 一种用于治疗ipf疾病的吸入制剂及其制备方法

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20090227

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK RS

DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20100301